Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Med Chem ; 2024 May 01.
Article in English | MEDLINE | ID: mdl-38691510

ABSTRACT

Metabotropic glutamate (mGlu) receptors play a key role in modulating most synapses in the brain. The mGlu7 receptors inhibit presynaptic neurotransmitter release and offer therapeutic possibilities for post-traumatic stress disorders or epilepsy. Screening campaigns provided mGlu7-specific allosteric modulators as the inhibitor XAP044 (Gee et al. J. Biol. Chem. 2014). In contrast to other mGlu receptor allosteric modulators, XAP044 does not bind in the transmembrane domain but to the extracellular domain of the mGlu7 receptor and not at the orthosteric site. Here, we identified the mode of action of XAP044, combining synthesis of derivatives, modeling and docking experiments, and mutagenesis. We propose a unique mode of action of these inhibitors, preventing the closure of the Venus flytrap agonist binding domain. While acting as a noncompetitive antagonist of L-AP4, XAP044 and derivatives act as apparent competitive antagonists of LSP4-2022. These data revealed more potent XAP044 analogues and new possibilities to target mGluRs.

2.
J Med Chem ; 67(2): 1314-1326, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38170918

ABSTRACT

Metabotropic glutamate (Glu) receptors (mGlu receptors) play a key role in modulating excitatory neurotransmission in the central nervous system (CNS). In this study, we report the structure-based design and pharmacological evaluation of densely functionalized, conformationally restricted glutamate analogue (1S,2S,3S)-2-((S)-amino(carboxy)methyl)-3-(carboxymethyl)cyclopropane-1-carboxylic acid (LBG30300). LBG30300 was synthesized in a stereocontrolled fashion in nine steps from a commercially available optically active epoxide. Functional characterization of all eight mGlu receptor subtypes showed that LBG30300 is a picomolar agonist at mGlu2 with excellent selectivity over mGlu3 and the other six mGlu receptor subtypes. Bioavailability studies on mice (IV administration) confirm CNS exposure, and an in silico study predicts a binding mode of LBG30300 which induces a flipping of Tyr144 to allow for a salt bridge interaction of the acetate group with Arg271. The Tyr144 residue now prevents Arg271 from interacting with Asp146, which is a residue of differentiation between mGlu2 and mGlu3 and thus could explain the observed subtype selectivity.


Subject(s)
Central Nervous System , Receptors, Metabotropic Glutamate , Mice , Animals , Central Nervous System/metabolism , Receptors, Metabotropic Glutamate/metabolism , Cyclopropanes/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Glutamates , Carboxylic Acids
3.
Elife ; 102021 12 06.
Article in English | MEDLINE | ID: mdl-34866572

ABSTRACT

G protein-coupled receptors (GPCRs) are among the most promising drug targets. They often form homo- and heterodimers with allosteric cross-talk between receptor entities, which contributes to fine-tuning of transmembrane signaling. Specifically controlling the activity of GPCR dimers with ligands is a good approach to clarify their physiological roles and validate them as drug targets. Here, we examined the mode of action of positive allosteric modulators (PAMs) that bind at the interface of the transmembrane domains of the heterodimeric GABAB receptor. Our site-directed mutagenesis results show that mutations of this interface impact the function of the three PAMs tested. The data support the inference that they act at the active interface between both transmembrane domains, the binding site involving residues of the TM6s of the GABAB1 and the GABAB2 subunit. Importantly, the agonist activity of these PAMs involves a key region in the central core of the GABAB2 transmembrane domain, which also controls the constitutive activity of the GABAB receptor. This region corresponds to the sodium ion binding site in class A GPCRs that controls the basal state of the receptors. Overall, these data reveal the possibility of developing allosteric compounds able to specifically modulate the activity of GPCR homo- and heterodimers by acting at their transmembrane interface.


Subject(s)
Receptors, GABA-B/genetics , Allosteric Regulation , HEK293 Cells , Humans , Ligands , Protein Domains , Receptors, GABA-B/metabolism
4.
Neuropharmacology ; 164: 107902, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31811873

ABSTRACT

Vesicular glutamate transporters (VGLUT1-3) mediate the uptake of glutamate into synaptic vesicles. VGLUTs are pivotal actors of excitatory transmission and of almost all brain functions. Their implication in various pathologies has been clearly documented. Despite their functional importance, the pharmacology of VGLUTs is limited to a few dyes such as Trypan Blue, Rose Bengal or Brilliant Yellow type. Here, we report the design and evaluation of new potent analogs based on Trypan Blue scaffold. Our best compound, named LSP5-2157, has an EC50 of 50 nM on glutamate vesicular uptake. Using a 3D homology model of VGLUT1 and docking experiments, we determined its putative binding subdomains within vesicular glutamate transporters and validated the structural requirement for VGLUT inhibition. To better estimate the specificity and potency of LSP5-2157, we also investigated its ability to block glutamatergic transmission in autaptic hippocampal cells. Neither glutamate receptors nor GABAergic transmission or transmission machinery were affected by LSP5-2157. Low doses of compound reversibly reduce glutamatergic neurotransmission in hippocampal autpases. LSP5-2157 had a low and depressing effect on synaptic efficacy in hippocampal slice. Furthermore, LSP5-2157 had no effect on NMDA-R- mediated fEPSP but reduce synaptic plasticity induced by 3 trains of 100 Hz. Finally, LSP5-2157 had the capacity to inhibit VGLUT3-dependent auditory synaptic transmission in the guinea pig cochlea. In this model, it abolished the compound action potential of auditory nerve at high concentration showing the limited permeation of LSP5-2157 in an in-vivo model. In summary, the new ligand LSP5-2157, has a high affinity and specificity for VGLUTs and shows some permeability in isolated neuron, tissue preparations or in vivo in the auditory system. These findings broaden the field of VGLUTs inhibitors and open the way to their use to assess glutamatergic functions in vitro and in vivo.


Subject(s)
Vesicular Glutamate Transport Proteins/antagonists & inhibitors , Action Potentials/drug effects , Animals , Cochlea/drug effects , Cochlear Nerve/drug effects , Excitatory Postsynaptic Potentials/drug effects , Guinea Pigs , Hippocampus/cytology , Hippocampus/drug effects , Mice , Mice, Inbred C57BL , Models, Molecular , Neurons/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Vesicular Glutamate Transport Proteins/metabolism
5.
Mol Pharmacol ; 96(2): 233-246, 2019 08.
Article in English | MEDLINE | ID: mdl-31189666

ABSTRACT

The orphan G-protein-coupled receptor (GPCR) GPR158 is expressed in the brain, where it is involved in the osteocalcin effect on cognitive processes, and at the periphery, where it may contribute to glaucoma and cancers. GPR158 forms a complex with RGS7-ß5, leading to the regulation of neighboring GPCR-induced Go protein activity. GPR158 also interacts with αo, although no canonical Go coupling has been reported. GPR158 displays three VCPWE motifs in its C-terminal domain that are putatively involved in G-protein regulation. Here, we addressed the scaffolding function of GPR158 and its VCPWE motifs on Go. We observed that GPR158 interacted with and stabilized the amount of RGS7-ß5 through a 50-residue region downstream of its transmembrane domain and upstream of the VCPWE motifs. We show that two VCPWE motifs are involved in αo binding. Using a Gαo-ßγ bioluminescence resonance energy transfer (BRET) sensor, we found that GPR158 decreases the BRET signal as observed upon G-protein activation; however, no constitutive activity of GPR158 could be detected through the measurement of various G-protein-mediated downstream responses. We propose that the effect of GPR158 on Go is unlikely due to a canonical activation of Go, but rather to the trapping of Gαo by the VCPWE motifs, possibly leading to its dissociation from ßγ Such action of GPR158 is expected to prolong the ßγ activity, as also observed with some activators of G-protein signaling. Taken together, our data revealed a complex functional scaffolding or signaling role for GPR158 controlling Go through an original mechanism.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , RGS Proteins/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Amino Acid Motifs , Binding Sites , Bioluminescence Resonance Energy Transfer Techniques , Gene Expression Regulation , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Protein Binding , Receptors, G-Protein-Coupled/genetics
6.
J Med Chem ; 61(5): 1969-1989, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29397723

ABSTRACT

A group III metabotropic glutamate (mGlu) receptor agonist (PCEP) was identified by virtual HTS. This orthosteric ligand is composed by an l-AP4-derived fragment that mimics glutamate and a chain that binds into a neighboring pocket, offering possibilities to improve affinity and selectivity. Herein we describe a series of derivatives where the distal chain is replaced by an aromatic or heteroaromatic group. Potent agonists were identified, including some with a mGlu4 subtype preference, e.g., 17m (LSP1-2111) and 16g (LSP4-2022). Molecular modeling suggests that aromatic functional groups may bind at either one of the two chloride regulatory sites. These agonists may thus be considered as particular bitopic/dualsteric ligands. 17m was shown to reduce GABAergic synaptic transmission at striatopallidal synapses. We now demonstrate its inhibitory effect at glutamatergic parallel fiber-Purkinje cell synapses in the cerebellar cortex. Although these ligands have physicochemical properties that are markedly different from typical CNS drugs, they hold significant therapeutic potential.


Subject(s)
Binding Sites , Receptors, Metabotropic Glutamate/agonists , Aminobutyrates/pharmacology , Animals , Glutamic Acid/chemistry , Humans , Ligands , Models, Molecular , Molecular Mimicry , Phosphinic Acids/pharmacology , Purkinje Cells/ultrastructure , Synapses/drug effects , Synaptic Transmission/drug effects
7.
FASEB J ; 29(10): 4174-88, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26116702

ABSTRACT

Metabotropic glutamate receptors (mGluRs) play key roles in the modulation of many synapses. Chloride (Cl(-)) is known to directly bind and regulate the function of different actors of neuronal activity, and several studies have pointed to the possible modulation of mGluRs by Cl(-). Herein, we demonstrate that Cl(-) behaves as a positive allosteric modulator of mGluRs. For example, whereas glutamate potency was 3.08 ± 0.33 µM on metabotropic glutamate (mGlu) 4 receptors in high-Cl(-) buffer, signaling activity was almost abolished in low Cl(-) in cell-based assays. Cl(-) potency was 78.6 ± 3.5 mM. Cl(-) possesses a high positive cooperativity with glutamate (Hill slope ≈6 on mGlu4), meaning that small variations in [Cl(-)] lead to large variations in glutamate action. Using molecular modeling and mutagenesis, we have identified 2 well-conserved Cl(-) binding pockets in the extracellular domain of mGluRs. Moreover, modeling of activity-dependent Cl(-) variations at GABAergic synapses suggests that these variations may be compatible with a dynamic modulation of the most sensitive mGluRs present in these synapses. Taken together, these data reveal a necessary role of Cl(-) for the glutamate activation of many mGluRs. Exploiting Cl(-) binding pockets may yield to the development of innovative regulators of mGluR activity.


Subject(s)
Chlorides/metabolism , Extracellular Space/metabolism , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation , Allosteric Site , Amino Acid Sequence , Animals , Binding Sites/genetics , Chlorides/pharmacology , HEK293 Cells , Humans , Models, Molecular , Mutation , Protein Binding , Protein Structure, Tertiary/drug effects , Rats , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/genetics , Sequence Homology, Amino Acid
8.
Org Biomol Chem ; 13(4): 1106-12, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25417857

ABSTRACT

A series of phosphinic glutamate derivatives (e.g.LSP1-2111) have been proven to be potent agonists of metabotropic glutamate (mGlu) receptors and shown promising in vivo activity. However, so far all were synthesized and tested as a mixture of two diastereomers whose absolute and relative configurations are not known. In this study, the stereomers were separated on a Crownpack CR(+) column and their absolute configuration was assessed by means of a diastereoselective synthesis. Both separated L-stereomers activated the mGlu4 receptor with EC50's of 0.72 and 4.4 µM for (1S,1'S)-and (1S,1'R)-LSP1-2111, respectively.


Subject(s)
Glutamic Acid/chemistry , Organophosphorus Compounds/chemistry , Glutamic Acid/pharmacology , HEK293 Cells , Humans , Models, Molecular , Molecular Conformation , Receptors, Metabotropic Glutamate/agonists
9.
FASEB J ; 26(4): 1682-93, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22223752

ABSTRACT

Metabotropic glutamate (mGlu) receptors are promising targets to treat numerous brain disorders. So far, allosteric modulators are the only subtype selective ligands, but pure agonists still have strong therapeutic potential. Here, we aimed at investigating the possibility of developing subtype-selective agonists by extending the glutamate-like structure to hit a nonconsensus binding area. We report the properties of the first mGlu4-selective orthosteric agonist, derived from a virtual screening hit, LSP4-2022 using cell-based assays with recombinant mGlu receptors [EC(50): 0.11 ± 0.02, 11.6 ± 1.9, 29.2 ± 4.2 µM (n>19) in calcium assays on mGlu4, mGlu7, and mGlu8 receptors, respectively, with no activity at the group I and -II mGlu receptors at 100 µM]. LSP4-2022 inhibits neurotransmission in cerebellar slices from wild-type but not mGlu4 receptor-knockout mice. In vivo, it possesses antiparkinsonian properties after central or systemic administration in a haloperidol-induced catalepsy test, revealing its ability to cross the blood-brain barrier. Site-directed mutagenesis and molecular modeling was used to identify the LSP4-2022 binding site, revealing interaction with both the glutamate binding site and a variable pocket responsible for selectivity. These data reveal new approaches for developing selective, hydrophilic, and brain-penetrant mGlu receptor agonists, offering new possibilities to design original bioactive compounds with therapeutic potential.


Subject(s)
Excitatory Amino Acid Agonists/chemistry , Excitatory Amino Acid Agonists/pharmacology , Ligands , Phosphinic Acids/chemistry , Phosphinic Acids/pharmacology , Receptors, Metabotropic Glutamate/agonists , Animals , Antiparkinson Agents/chemistry , Antiparkinson Agents/metabolism , Antiparkinson Agents/pharmacology , Binding Sites , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Molecular Structure , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Phosphinic Acids/metabolism , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Synaptic Transmission/drug effects
10.
Nat Chem Biol ; 6(8): 587-94, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20622858

ABSTRACT

G protein-coupled receptor (GPCR) oligomers have been proposed to play critical roles in cell signaling, but confirmation of their existence in a native context remains elusive, as no direct interactions between receptors have been reported. To demonstrate their presence in native tissues, we developed a time-resolved FRET strategy that is based on receptor labeling with selective fluorescent ligands. Specific FRET signals were observed with four different receptors expressed in cell lines, consistent with their dimeric or oligomeric nature in these transfected cells. More notably, the comparison between FRET signals measured with sets of fluorescent agonists and antagonists was consistent with an asymmetric relationship of the two protomers in an activated GPCR dimer. Finally, we applied the strategy to native tissues and succeeded in demonstrating the presence of oxytocin receptor dimers and/or oligomers in mammary gland.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Oligopeptides/chemistry , Receptors, G-Protein-Coupled/metabolism , Algorithms , Animals , Antidiuretic Hormone Receptor Antagonists , COS Cells , Cell Line , Chlorocebus aethiops , Dimerization , Dopamine D2 Receptor Antagonists , Female , Fluorescent Dyes , Ligands , Mammary Glands, Animal/metabolism , Models, Molecular , Oligopeptides/metabolism , Radioligand Assay , Rats , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Oxytocin/agonists , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism , Receptors, Vasopressin/agonists , Receptors, Vasopressin/metabolism
11.
J Med Chem ; 53(7): 2797-813, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-20218620

ABSTRACT

(R)-PCEP (3-amino-3-carboxypropyl-2'-carboxyethyl phosphinic acid, 1), a new metabotropic glutamate receptor 4 (mGlu4R) agonist, was discovered in a previously reported virtual screening. The (S)-enantiomer and a series of derivatives were synthesized and tested on recombinant mGlu4 receptors. A large number of derivatives activated this receptor but was not able to discriminate between mGlu4 and mGlu8 receptors. The most potent ones 6 and 12 displayed an EC(50) of 1.0 +/- 0.2 microM at mGlu4R. Interestingly these agonists with longer alkyl chains revealed a new binding pocket adjacent to the glutamate binding site, which is lined with residues that differ among the mGluR subtypes and that will allow the design of more selective compounds. Additionally 6 was able to activate mGlu7 receptor with an EC(50) of 43 +/- 16 microM and is thus significantly more potent than L-AP4 (EC(50) of 249 +/- 106 microM).


Subject(s)
Drug Design , Drug Evaluation, Preclinical/methods , Receptors, Metabotropic Glutamate/agonists , User-Computer Interface , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Dose-Response Relationship, Drug , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Phosphinic Acids/chemical synthesis , Phosphinic Acids/chemistry , Phosphinic Acids/metabolism , Phosphinic Acids/pharmacology , Rats , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/metabolism , Stereoisomerism , Structure-Activity Relationship
12.
J Med Chem ; 50(15): 3585-95, 2007 Jul 26.
Article in English | MEDLINE | ID: mdl-17602546

ABSTRACT

Stereoisomers of 1-amino-2-phosphonomethylcyclopropanecarboxylic acid (APCPr), conformationally restricted analogues of L-AP4 (2-amino-4-phosphonobutyric acid), have been prepared and evaluated at recombinant group III metabotropic glutamate receptors. They activate these receptors over a broad range of potencies. The most potent isomer (1S,2R)-APCPr displays a similar pharmacological profile as that of L-AP4 (EC50 0.72, 1.95, >500, 0.34 microM at mGlu4, 6, 7, 8 receptors, respectively, and no effect at group I/II mGluRs). It was characterized on native receptors located in the basal ganglia (BG) where it induced a robust and reversible inhibition of synaptic transmission. It was tested in vivo in haloperidol-induced catalepsy, a model of Parkinsonian akinesia, by direct infusion in the globus pallidus of the BG. At a dose of 0.5 nmol/microL, catalepsy was significantly antagonized. This study reveals that (1S,2R)-APCPr is a potent group III mGluR agonist and confirms that these receptors may be considered as a therapeutic target in the Parkinson's disease.


Subject(s)
Amino Acids/chemical synthesis , Antiparkinson Agents/chemical synthesis , Organophosphonates/chemical synthesis , Receptors, Metabotropic Glutamate/agonists , Amino Acids/chemistry , Amino Acids/pharmacology , Animals , Antiparkinson Agents/chemistry , Antiparkinson Agents/pharmacology , Basal Ganglia/drug effects , Basal Ganglia/physiology , Catalepsy/chemically induced , Catalepsy/drug therapy , Cell Line , Haloperidol , Humans , In Vitro Techniques , Injections , Male , Models, Molecular , Molecular Conformation , Organophosphonates/chemistry , Organophosphonates/pharmacology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/drug effects , Stereoisomerism , Structure-Activity Relationship , Synaptic Transmission/drug effects
13.
Biochem Biophys Res Commun ; 349(2): 471-9, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16945339

ABSTRACT

The seven-transmembrane receptor Smoothened (Smo) transduces the signal initiated by Hedgehog (Hh) morphogen binding to the receptor Patched (Ptc). We have reinvestigated the pharmacological properties of reference molecules acting on the Hh pathway using various Hh responses and a novel functional assay based on the coexpression of Smo with the alpha subunit of the G15 protein in HEK293 cells. The measurement of inositol phosphate (IP) accumulation shows that Smo has constitutive activity, a response blocked by Ptc which indicates a functional Hh receptor complex. Interestingly, the antagonists cyclopamine, Cur61414, and SANT-1 display inverse agonist properties and the agonist SAG has no effect at the Smo-induced IP response, but converts Ptc-mediated inactive forms of Smo into active ones. An oncogenic Smo mutant does not mediate an increase in IP response, presumably reflecting its inability to reach the cell membrane. These studies identify novel properties of molecules displaying potential interest in the treatment of various cancers and brain diseases, and demonstrate that Smo is capable of signaling through G15.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Genes, Tumor Suppressor/physiology , RNA-Binding Proteins/physiology , Receptors, G-Protein-Coupled/physiology , Trans-Activators/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Line , Cell Membrane/metabolism , Dioxoles/pharmacology , Hedgehog Proteins , Humans , Mice , Neoplasms/metabolism , Piperazines/pharmacology , Pyrazoles/pharmacology , Signal Transduction , Smoothened Receptor , Stem Cells/metabolism , Veratrum Alkaloids/pharmacology
14.
J Med Chem ; 48(7): 2534-47, 2005 Apr 07.
Article in English | MEDLINE | ID: mdl-15801843

ABSTRACT

The "receiver operating characteristic" (ROC) curve method is a well-recognized metric used as an objective way to evaluate the ability of a given test to discriminate between two populations. This facilitates decision-making in a plethora of fields in which a wrong judgment may have serious consequences including clinical diagnosis, public safety, travel security, and economic strategies. When virtual screening is used to speed-up the drug discovery process in pharmaceutical research, taking the right decision upon selecting or discarding a molecule prior to in vitro evaluation is of paramount importance. Characterizing both the ability of a virtual screening workflow to select active molecules and the ability to discard inactive ones, the ROC curve approach is well suited for this critical decision gate. As a case study, the first virtual screening workflow focused on metabotropic glutamate receptor subtype 4 (mGlu4R) agonists is reported here. Six compounds out of 38 selected and tested in vitro were shown to have agonist activity on this target of therapeutic interest.


Subject(s)
Drug Design , Quantitative Structure-Activity Relationship , ROC Curve , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/chemistry , Binding Sites , Databases, Factual , Models, Molecular
15.
J Biol Chem ; 279(16): 15824-30, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-14736871

ABSTRACT

The gamma-aminobutyric acid type B (GABAB) receptor is an allosteric complex made of two subunits, GABAB1 (GB1) and GABAB2 (GB2). Both subunits are composed of an extracellular Venus flytrap domain (VFT) and a heptahelical domain (HD). GB1 binds GABA, and GB2 plays a major role in G-protein activation as well as in the high agonist affinity state of GB1. How agonist affinity in GB1 is regulated in the receptor remains unknown. Here, we demonstrate that GB2 VFT is a major molecular determinant involved in this control. We show that isolated versions of GB1 and GB2 VFTs in the absence of the HD and C-terminal tail can form hetero-oligomers as shown by time-resolved fluorescence resonance energy transfer (based on HTRF technology). GB2 VFT and its association with GB1 VFT controlled agonist affinity in GB1 in two ways. First, GB2 VFT exerted a direct action on GB1 VFT, as it slightly increased agonist affinity in isolated GB1 VFT. Second and most importantly, GB2 VFT prevented inhibitory interaction between the two main domains (VFT and HD) of GB1. According to this model, we propose that GB1 HD prevents the possible natural closure of GB1 VFT. In contrast, GB2 VFT facilitates this closure. Finally, such inhibitory contacts between HD and VFT in GB1 could be similar to those important to maintain the inactive state of the receptor.


Subject(s)
Receptors, GABA-B/chemistry , Allosteric Regulation , Binding Sites , Cell Line , GABA Agonists/pharmacology , GABA-B Receptor Agonists , Humans , Models, Molecular , Protein Subunits , Signal Transduction , Structure-Activity Relationship
16.
J Biol Chem ; 278(37): 35063-70, 2003 Sep 12.
Article in English | MEDLINE | ID: mdl-12829705

ABSTRACT

Heptahelical receptor coupling selectivity to G-proteins is controlled by a large contact area that involves several portions of the receptor and each subunit of the G-protein. In the G-protein alpha subunit, the C-terminal 5 residues, the N terminus, and the alpha N-beta 1 and alpha 4-alpha 5 loops play important roles. On the receptor side, both the second and third (i2 and i3) intracellular loops as well as the C-terminal tail probably contact these different regions of the G-protein. It is now accepted that the C terminus of the alpha subunit binds in a cavity formed by the i2 and i3 loops. Among the various G-protein-coupled receptors (GPCRs), class III receptors that include metabotropic glutamate (mGlu) receptors greatly differ from the rhodopsin-like GPCRs, but the contact zone between these receptors and the G-protein is less understood. The C terminus of the alpha subunit has been shown to play a pivotal role in the selective recognition of class III GPCRs. Indeed, the mGlu2 and mGlu4 and -8 receptors can discriminate between alpha subunits that differ at the level of their C-terminal end only (such as Gqo and Gqz). Here, we examine the role of the i2 loop of mGluRs in the selective recognition of this region of the alpha subunit. To that aim, we analyzed the coupling properties of mGlu2 and mGlu4 or -8 receptors and chimeras containing the i2 loop of the converse receptor to G-protein alpha subunits that only differ by their C termini (Gqo,Gqz, and their point mutants). Our data demonstrate that the central portion of the i2 loop is responsible for the selective recognition of the C-terminal end of the alpha subunit, especially the residue on position -4. These data are consistent with the proposal that the C-terminal end of the G-protein alpha subunit interacts with residues in a cavity formed by the i2 and i3 loops in class III GPCRs, as reported for class I GPCRs.


Subject(s)
GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Receptors, Metabotropic Glutamate/chemistry , Amino Acid Sequence , Binding Sites , Cell Line , GTP-Binding Proteins/genetics , Humans , Inositol Phosphates/metabolism , Kidney , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
17.
Proc Natl Acad Sci U S A ; 99(17): 11097-102, 2002 Aug 20.
Article in English | MEDLINE | ID: mdl-12151600

ABSTRACT

Ca2+, pheromones, sweet taste compounds, and the main neurotransmitters glutamate and gamma-aminobutyric acid activate G protein-coupled receptors (GPCRs) that constitute the GPCR family 3. These receptors are dimers, and each subunit has a large extracellular domain called a Venus flytrap module (VFTM), where agonists bind. This module is connected to a heptahelical domain that activates G proteins. Recently, the structure of the dimer of mGlu1 VFTMs revealed two important conformational changes resulting from glutamate binding. First, agonists can stabilize a closed state of at least one VFTM in the dimer. Second, the relative orientation of the two VFTMs in the dimer is different in the presence of glutamate, such that their C-terminal ends (which are connected to the G protein-activating heptahelical domain) become closer by more than 20 A. This latter change in orientation has been proposed to play a key role in receptor activation. To elucidate the respective role of VFTM closure and the change in orientation of the VFTMs in family 3 GPCR activation, we analyzed the mechanism of action of the mGlu8 receptor antagonists ACPT-II and MAP4. Molecular modeling studies suggest that these two compounds prevent the closure of the mGlu8 VFTM because of ionic and steric hindrance, respectively. We show here that the replacement of the residues responsible for these hindrances (Asp-309 and Tyr-227, respectively) by Ala allows ACPT-II or MAP4 to fully activate the receptors. These data are consistent with the requirement of the VFTM closure for family 3 GPCR activation.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/physiology , Amino Acid Sequence , Amino Acid Substitution , Cell Line , Dimerization , GTP-Binding Proteins/physiology , Glutamic Acid/physiology , Humans , Kidney , Kinetics , Ligands , Mutagenesis, Site-Directed , Protein Conformation , Receptors, Metabotropic Glutamate/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...